WO2007059000A2 - Modulation de l'angiogenese au moyen de fragments de peptide a-beta - Google Patents

Modulation de l'angiogenese au moyen de fragments de peptide a-beta Download PDF

Info

Publication number
WO2007059000A2
WO2007059000A2 PCT/US2006/043921 US2006043921W WO2007059000A2 WO 2007059000 A2 WO2007059000 A2 WO 2007059000A2 US 2006043921 W US2006043921 W US 2006043921W WO 2007059000 A2 WO2007059000 A2 WO 2007059000A2
Authority
WO
WIPO (PCT)
Prior art keywords
fragment
peptide
angiogenic
peptide fragment
angiogenesis
Prior art date
Application number
PCT/US2006/043921
Other languages
English (en)
Other versions
WO2007059000A3 (fr
Inventor
Daniel Paris
Michael J. Mullan
Original Assignee
Roskamp Research, Llc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Roskamp Research, Llc filed Critical Roskamp Research, Llc
Priority to EP06837407A priority Critical patent/EP1951750A4/fr
Priority to CA002619455A priority patent/CA2619455A1/fr
Priority to AU2006315686A priority patent/AU2006315686A1/en
Priority to JP2008540237A priority patent/JP2009516654A/ja
Publication of WO2007059000A2 publication Critical patent/WO2007059000A2/fr
Publication of WO2007059000A3 publication Critical patent/WO2007059000A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4711Alzheimer's disease; Amyloid plaque core protein
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof

Definitions

  • the present invention is related to compositions and methods for treating diseases and pathological conditions or processes mediated by pathological angiogenesis by administering biologically active fragments of full length A ⁇ peptides to a patient suffering from such diseases, conditions, or processes.
  • AD Alzheimer's disease
  • APP amyloid precursor protein
  • the primary protein component of senile plaques is beta/A4 amyloid, a 42-43 amino acid peptide.
  • Vascular pathology is the norm in advanced cases of AD, with cerebral amyloid angiopathy (CAA) being one of the most common abnormalities detected at autopsy (Ellis, et al. Neurology 46:1592-1596 (1996)). Certain vascular lesions, such as microvascular degeneration affecting the cerebral endothelium and periventricular white matter lesions, are evident in most AD cases (Ellis, et al. Neurology 46:1592- 1596 (1996); Kalaria, Ann. N.Y. Acad. Sci. 893:113-125 (1999)).
  • CAA cerebral amyloid angiopathy
  • AD brain microvessels and capillaries have been observed in AD brain microvessels and capillaries; in particular, terminal arterioles frequently have focal constriction and smooth muscle cells with an irregular shape and arrangement (Hashimura et al. Jpn. J. Psychiatry Neurol. 45:661-665 (1991)).
  • Capillaries in AD brain typically show an abnormal abluminal surface with irregular constriction and dilatation along their paths (Kimura et al. Jpn. J. Psychiatry Neurol. 45:671-676 (1991)).
  • a ⁇ peptides are known to form fibrillar deposits around blood vessels, leading to cerebral amyloid angiopathy (CAA) (Pardridge, et al. 1987 J. Neurochem. 49, 1394- 401; Jellinger K.A., Attems J. 2005 J. Neurol. Sci. 229-230, 37-41).
  • CAA cerebral amyloid angiopathy
  • vascular functional impairments and reduced blood flow are characteristic features of the AD brain (Nicoll, et al. 2004 Neurobiol. Aging. 25, 589- 97 and 603-4; Paris, et al. 2004 Brain Res. 999, 53-61; Beckmann, et al. 2003 J. Neurosci. 23, 8453-9; Farkasm, et al.
  • Angiogenesis is inhibited by A ⁇ peptides in multiple different in-vitro and in- vivo assays (Paris, et al. 2004 Angiogenesis. 7, 75-85).
  • a ⁇ o and A ⁇ i -42 can dose dependency inhibit capillary tube formation by human brain microvascular endothelial cells when plated on Matrigel, and can promote capillary degeneration at high doses.
  • Mutants of the full-length A ⁇ peptide, including 1 or 2 amino acid substitutions, were also found to be biologically active anti-angiogenics. However at low doses, A ⁇ appears to be pro-angiogenic (Paris, et al. 2004 Angiogenesis. 7, 75-85; Cantara, et al. 2004F.A.S.E.B. J. 18, 1943-5).
  • a ⁇ peptides are useful as anti-angiogenic agents. These anti-angiogenic A ⁇ peptide fragments may be used to treat pathological conditions mediated by undesired and/or uncontrolled angiogenesis (characterized as "angiogenic diseases"), as described further herein.
  • the present invention provides a variety of anti- angiogenic A ⁇ peptide fragments as well as compositions which include one or more such fragments.
  • the biologically active A ⁇ peptide fragment may be 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38 or 39 amino acids in length.
  • the anti-angiogenic A ⁇ peptide fragment is the A ⁇ i -2 8 peptide fragment, the A ⁇ io-35 peptide fragment, the A ⁇ i 2-28 peptide fragment, the Ap 1 3-20 peptide fragment, or other biologically active fragments or variants or homologs thereof.
  • the anti-angiogenic A ⁇ peptide fragment is Ap 12-28 and contains the amino acid sequence HHQKIvVFF, or biologically active fragments, variants or homologs thereof.
  • the anti-angiogenic A ⁇ peptide fragment is A ⁇ i3 -20 or the amino acid sequence HHGKLVFF, or biologically active variants or homologs thereof.
  • the variants may include, for example, amino acid substitutions.
  • the present invention is a pharmaceutical composition
  • a pharmaceutical composition comprising an anti-angiogenic A ⁇ peptide fragment and one or more pharmaceutically acceptable carriers, diluents, or excipients.
  • the present invention provides a method for treating a disease or disorder mediated by pathological angiogenesis by administering to a subject in need thereof an effective amount of a biologically active Ap peptide fragment, wherein the fragment is between 8 and 39 amino acids in length.
  • the anti- angiogenic Ap peptide fragment is optionally administered in combination or alternation with one or more therapeutic agents.
  • the subject may be, for example, a mammal such as a human.
  • the present invention is a method for treating cancer by administering to a subject in need thereof an effective amount of a biologically active Ap peptide fragment, optionally, in combination or alternation with one or more chemotherapeutic agents.
  • the present invention is a method of treating cancer by administering to a subject in need thereof an effective amount of a APi 2-38 peptide fragment containing the amino acid sequence HHQKLVFF or biologically active fragments, variants or homologs thereof.
  • the method of treating cancer involves administering to a subject in need thereof an effective amount of APi 3-20 peptide fragment or the amino acid sequence HHQKLVFF or biologically active variants or homologs thereof.
  • the biologically active A ⁇ peptide fragment can be administered by any suitable means including, but not limited, to oral, parenteral, intravenous, intraarterial, pulmonary, mucosal, topical, transdermal, subcuteaneous, intramuscular, intrathecal or intraperitoneal administration.
  • a third aspect of the present invention provides diagnostic methods and kits for detection and measurement of anti-angiogenic A ⁇ peptide fragment activity in biological fluids and tissues.
  • a fourth aspect of the present invention provides diagnostic methods and kits to screen for compounds that are potentially therapeutic in treatment of Alzheimer's disease by interfering with the anti-angiogenic effect of the A ⁇ peptide fragment.
  • Another aspect of the invention are uses of the peptide fragments in
  • Figure 1 is a graph of the total length of capillary tubes expressed as a percentage of control treatment for 0, 1, 5 and 10 ⁇ M doses of various A ⁇ peptide fragments as described in Example 8.
  • Figures 2A and 2B are charts of the cellular proliferation and cellular adhesion of HUVEC samples, expressed as a percentage of the control, after incubation with various A ⁇ peptide fragments as described in Example 9.
  • Figure 3 is a chart of the total length of capillary tubes expressed as a percentage of control treatment versus treatment with heparin (0.5 or 1 mg/ml), A ⁇ l-42 peptide, A ⁇ + heparin (500 ⁇ g/ml) and A ⁇ + heparin (lmg/ml) as described in Example 10.
  • Figure 4 is a graph of the total length of capillary tubes expressed as a percentage of control treatment for 0, 1, 5 and 10 ⁇ M doses of A ⁇ i. 28 , A ⁇ i -28 GGQGL and A ⁇ i- 28 AAQAL as described in Example 11.
  • Figure 5 provides photographs (at 4X magnification) of capillaries tubes formed following incubation with A ⁇ peptide fragments as described in Example 11.
  • Figure 6 is a graph of the total length of capillary tubes expressed as a percentage of control treatment for 0, 1, 5 and 10 ⁇ M doses of the peptides HHHQKLVFF, VHHQKLVII, and VHHQKLVKK as described in Example 12.
  • Figure 7 is a chart of the Angiogenic Index (AI) for the rat corneal micropocket assay in response to 200ng VEGF, VEGF + 0.5 ⁇ g A ⁇ i 2-28 , VEGF + 2.5 ⁇ g A ⁇ ]2-28 and VEGF + 5.0 ⁇ g A ⁇ 12-28 as described in Example 13.
  • AI Angiogenic Index
  • Figure 8 is a chart of the Angiogenic Index (AI) for the rat corneal micropocket assay in response to VEGF, 5ug A ⁇ i -2 8 GGQGL, and 0.5ug, 2.5ug and 5ug of A ⁇ 12-28 and HHH-peptide (HHHQKLVFF), as described in Example 14.
  • AI Angiogenic Index
  • Figure 9 provides representative photographs of rat corneal micropockets following a seven day incubation as described in Example 14, including a VEGF control and 0.5 ⁇ g, 2.5 ⁇ g and 5.0 ⁇ g of A ⁇ 12-28 .
  • Figure 10 is a listing of sequences of A-Beta peptides and APP as well as the nucleic acids encoding them, as described in U.S. Patent Publication No. 2003/0077261 to Paris et al.
  • Anti-angiogenic therapy is an attractive approach for inhibition of tumor progression, as tumors depend upon an adequate blood supply for growth. It is disclosed herein that short peptides derived from the A ⁇ sequence inhibit angiogenesis, and can be used for anti-cancer therapy.
  • anti-angiogenic A ⁇ peptide fragments that can be used to treat pathological conditions mediated by undesired and/or uncontrolled or pathological angiogenesis.
  • HHQKLVFF anti-angiogenic motif
  • HHQKLVFF anti-angiogenic motif
  • a ⁇ peptide fragment refers to an anti-angiogenic fragment of a full length A ⁇ peptides (e.g., A ⁇ i -40 , A ⁇ 42 , A ⁇ 1-4 3) and includes A ⁇ peptide fragment variants, homologs (such as mammalian orthologs) and isoforms, unless otherwise noted.
  • the term also includes fragments with substitutions of one or more equivalent amino acids, or non-natural amino acids.
  • the A ⁇ peptide fragment is at least one amino acid less in number than the total number of amino acids found in the full-length A ⁇ peptide.
  • Full length A ⁇ peptides are derived from proteolytic processing of one or more isoforms of the amyloid precursor protein (APP), a transmembrane glycoprotein (Kang, J. et al. Nature (Lond.). (1987) 325: 733-736).
  • the 39-43-amino acid-long A ⁇ peptide amino acid sequence begins in the ectodomain of APP and extends into the transmembrane region.
  • a ⁇ is formed after sequential cleavage of APP by the ⁇ - and ⁇ -secretases.
  • a ⁇ 1-42 and A ⁇ i -43 forms are specifically found in all kinds of AD plaques, indicating that those forms are critically important in AD pathology.
  • the A ⁇ peptide fragment is at least one amino acid less in number than the total number of amino acids found in the full length A ⁇ i_ 4 o peptide.
  • the A ⁇ i- 4 o peptide fragment consists of, for example, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38 or 39 amino acids.
  • the A ⁇ peptide fragment is at least one amino acid less in number than the total number of amino acids found in the full length A ⁇ i. 42 peptide.
  • the A ⁇ i -42 fragment consists of, for example, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40 or 41 amino acids.
  • the A ⁇ peptide fragment is at least one amino acid less in number than the total number of amino acids found in the full length A ⁇ i_ 43 peptide.
  • the A ⁇ i -43 fragment consists of, for example, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 41, or 42 amino acids.
  • the fragment consists of, for example, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28 or more amino acid residues, and includes the sequence HHQKLVFF.
  • one or more of the following biologically active A ⁇ peptide fragments may be used to treat diseases or disorders associated with unwanted or pathological angiogenesis: the A ⁇ ]-2 8 peptide, the Ap I0-35 peptide, the A ⁇ i 2-2 g peptide, the A ⁇ I3-20 peptide, or biologically active fragments or variants thereof.
  • the anti-angiogenic A ⁇ peptide fragment preferably contains the HHQK proteoglycan binding region, since fragments without that sequence (A ⁇ 2 s -35 , A ⁇ i 7-2 g, and A ⁇ 34-42 ) were not active, suggesting that the heparin binding motif HHQK is required to mediate the anti-angiogenic activity of A ⁇ .
  • the A ⁇ io-i ⁇ fragment was inactive even though it contains the HHQK sequence, suggesting that the HHQK proteoglycan binding motif is not sufficient to inhibit angiogenesis and that other neighboring residues are required.
  • the LVFF sequence immediately following the HHQK domain is also required for inhibition of angiogenesis.
  • preferred A ⁇ peptide fragments contain the amino acid sequence HHQKLVFF.
  • the fragment consists of, for example, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 25, 36, 37, 38 or more amino acid residues, and includes the sequence HHQKLVFF.
  • Such fragments may include one or more (e.g. 2, 3 or 4) substitutions of equivalent amino acids, including, e.g., non-natural amino acids.
  • the A ⁇ peptide fragment is a A ⁇ i 2-2 8 peptide containing the amino acid sequence HHQKLVFF, or a biologically active fragment or variant thereof.
  • the A ⁇ peptide fragment is a Ap 13-2O peptide fragment or the amino acid sequence HHQKLVFF, or a biologically active fragment or valiant thereof.
  • the A ⁇ peptide fragment is, e.g., a 10, 20, 30, or 40 amino acid fragment of the A ⁇ peptide.
  • the peptide fragments are obtained, for example, by chemical synthesis, or are recombinantly produced by host cells.
  • variant and homologous are also used interchangeably.
  • “Variant” or “homologous” peptide fragments will be understood to designate those containing, in relation to the native polypeptide sequence, modifications such as deletion, addition, or substitution of at least one amino acid, truncation, extension, or the addition of chimeric heterologous polypeptides.
  • “variant” or “homologous” peptide fragments can contain a mutation or post-translational modifications.
  • polypeptides or peptide fragments those whose amino acid sequence exhibits 80.0% to 99.9% (inclusive) identity to the native polypeptide sequence are preferred. These percentages are purely statistical and differences between two peptide sequences can be distributed randomly and over the entire sequence length.
  • "Variant" or “homologous" polypeptide sequences exhibiting a percentage identity with the polypeptides of the present invention can, alternatively, have 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99 percent identity with the polypeptide sequences of the instant invention.
  • the expression equivalent amino acid is intended here to designate any amino acid capable of being substituted for one of the amino acids in the basic structure without, however, essentially modifying the biological activities of the corresponding peptides and as provided below.
  • HHQKLVFF motif region of A ⁇ while potentially retaining the substitution antiangiogenic properties of the peptide.
  • HHQEXVFF Several substitutions could be made to HHQKLVFF (motif) region of A ⁇ while potentially retaining the substitution antiangiogenic properties of the peptide. Specifically, the following expression indicates such equivalent substitutions for HHQEXVFF:
  • the A ⁇ peptide fragment consists of or comprises one of the peptide sequences listed in Table 1, with optional equivalent amino acid substitutions.
  • the subject invention also provides biologically active peptide fragments capable of eliciting an immune response.
  • the immune response can provide components (either antibodies or components of the cellular immune response (e.g., B-cells, helper, cytotoxic, and/or suppressor T-cells) reactive with the peptide fragment.
  • Fragments as described herein, can be obtained by cleaving a polypeptide with a proteolytic enzyme (such as trypsin, chymotrypsin, or collagenase) or with a chemical reagent, such as cyanogen bromide (CNBr).
  • a proteolytic enzyme such as trypsin, chymotrypsin, or collagenase
  • a chemical reagent such as cyanogen bromide (CNBr).
  • polypeptide fragments can be generated in a highly acidic environment, for example at pH 2.5.
  • Such polypeptide fragments may be also prepared by chemical synthesis or using hosts transformed with an expression vector containing nucleic acids encoding polypeptide fragments. The transformed host cells contain a nucleic acid and are cultured according to well-known methods; thus, expression of these fragments is possible, under the control of appropriate elements for regulation and/or expression.
  • the peptides can be modified by variation in the splicing of transcriptional products of the A ⁇ gene, genetic recombination, or by chemical synthesis. Such peptides can contain at least one modification in relation to the polypeptide sequence being modified. These modifications can include the addition, substitution, deletion of amino acids contained within the polypeptides.
  • amino acids of one class are replaced with another amino acid of the same type
  • the class of nonpolar amino acids include Ala, VaI, Leu, Ue, Pro, Met, Phe, GIy and Trp
  • the class of uncharged polar amino acids include Ser, Thr, Cys, Tyr, Asn, and GIn
  • the class of acidic amino acids includes Asp and GIu
  • the class of basic amino acids includes Lys, Arg, and His.
  • non-natural amino acids for example in the D form
  • amino acid analogs such as sulfur-containing forms of amino acids
  • Alternative means for increasing the life of polypeptides can also be used.
  • peptide fragments can be recombinantly modified to include elements that increase the plasma, or serum half-life. These elements include, and are not limited to, antibody constant regions (see for example, U.S. Patent No. 5,565,335, hereby incorporated by reference in its entirety, including all references cited therein), or other elements such as those disclosed in U.S. Patent Nos.
  • polypeptide fragments disclosed may further contain linkers that facilitate the attachment of the fragments to a carrier molecule for delivery or diagnostic purposes.
  • linkers can also be used to attach fragments to solid support matrices for use in affinity purification protocols, hi one embodiment, the linkers specifically exclude where the fragment is a subsequence of another peptide, polypeptide, or protein as identified in a search of protein sequence databases. In other words, the non-identical portions of the other peptide, polypeptide, of protein is not considered to be a "linker” in this aspect.
  • linkers suitable for the practice of the invention include chemical linkers (such as those sold by Pierce, Rockford, 111.), peptides that allow for the connection of the immunogenic fragment to a carrier molecule (see, for example, linkers disclosed in U.S. Patent Nos.
  • the linkers can be up to 50 amino acids in length, up to 40 amino acids in length, up to 30 amino acids in length, up to 20 amino acids in length, up to 10 amino acids in length, or up to 5 amino acids in length.
  • the peptides may be expressed as a fusion, or chimeric protein product ( joined via a peptide bond to a heterologous protein sequence (e.g., a different protein)).
  • a chimeric product can be made by ligating the appropriate nucleic acid sequences encoding the desired amino acid sequences to each other by methods known in the art, in the proper coding frame, and expressing the chimeric product by methods commonly known in the art (see, for example, U.S. Patent No. 6,342,362, hereby incorporated by reference in its entirety; Altendorf, et al.
  • a chimeric product may be made by protein synthetic techniques, e.g., by use of a peptide synthesizer. Fusion peptides can comprise polypeptides and one or more protein transduction domains, as described above. Such fusion peptides are particularly useful for delivering the cargo polypeptide through the cell membrane.
  • the amount of A ⁇ peptide fragment activity within a tissue is useful in treating a variety of angiogenic diseases, such as cancers, tumors, and/or malignancies.
  • the amount of A ⁇ peptide fragment activity can be increased within a tissue by directly administering the A ⁇ peptide fragment to a patient suffering from an angiogenic disease (such as exogenous delivery of the A ⁇ peptide fragment) or by indirect or genetic means (such as delivery of a polynucleotide encoding the A ⁇ peptide fragment or upregulating the endogenous A ⁇ peptide fragment activity).
  • Non-limiting examples of such cancers, tumors, and/or malignancies that can be treated using the methods of the invention include prostate cancer, breast cancer, melanoma, chronic myelogenous leukemia, cervical cancer, adenocarcinomas, lymphoblastic leukemia, colorectal cancer, and lung carcinoma.
  • the peptide fragments or nucleic acids encoding them can be used in screening, or aiding in the diagnosis of, an individual suspected of having an angiogenic or angiogenesis-mediated disease.
  • the peptide fragments disclosed herein and nucleic acids encoding them can be used to detect the A ⁇ peptide in hybridization assays by the use of complementary sequences.
  • the presence of a significantly increased amount of A ⁇ peptide fragment is associated with an indication of Alzheimer's disease.
  • the presence of a significantly decreased amount of A ⁇ peptide is associated with an indication of an angiogenic disease, such as a malignancy or cancer.
  • a ⁇ gene product can be detected by well-known methodologies including, and not limited to, Western blots, enzyme linked immunoassays (ELISAs), radioimmunoassays (RIAs), Northern blots, Southern blots, PCR-based assays, or other assays for the quantification of gene product known to the skilled artisan.
  • This information in conjunction with other information available to the skilled practitioner, assists in making a diagnosis.
  • the subject invention concerns a method of inhibiting angiogenesis in a patient in need of anti-angiogenesis therapy by administration of biologically active A ⁇ peptide fragment to the patient.
  • a treatment for a pathological condition selected from the group consisting of cancer, arthritis, atherosclerosis, psoriasis, macular degeneration, and diabetic retinopathy by administering to the patient a therapeutically effective amount of an A ⁇ peptide fragment.
  • biologically active variants of the A ⁇ peptide fragments are utilized, wherein the variants have a substitution at the 21 amino acid position, or the 22 amino acid position, or 23 amino acid position, or combinations thereof.
  • the substitution(s) is a conservative substitution which does not materially alter the biological activity of the polypeptide.
  • PTDs protein transduction domains
  • examples of PTDs include the Drosophila homeotic transcription protein antennapedia (Antp), the herpes simples virus structural protein VP22, and the human immuno-deficiency virus 1 (HIV-I) transcriptional activator Tat protein.
  • recombinant cells can be administered to a patient, wherein the recombinant cells have been genetically modified to express A ⁇ peptide fragments disclosed herein.
  • the method of angiogenesis inhibition provided can be used to treat a patient suffering from cancer, or as a cancer preventative.
  • the method of tumor inhibition provided can be used to treat patients suffering from a variety of cancers including, but not limited, to cancer of the breast, prostate, melanoma, chronic myelogenous leukemia, cervical cancer, adenocarcinoma, lymphoblastic leukemia, colorectal cancer, and lung carcinoma.
  • various other anticancer or anti-rumor compounds such as cytotoxic agents, can be administered in conjunction with A ⁇ peptide fragments. Nucleotide sequences encoding A ⁇ fragments
  • the subject invention provides isolated and/or purified nucleotide sequences comprising a polynucleotide sequence encoding the amino acid sequence of the peptide fragments disclosed herein.
  • isolated nucleic acid molecules comprising polynucleotides encoding the A ⁇ peptide fragments.
  • One aspect of the invention provides isolated nucleic acid molecules comprising polynucleotides having a nucleotide sequence selected from the group consisting of: (a) a nucleotide sequence encoding any of the amino acid sequences of the polypeptides described herein including in Table 1; and (b) a nucleotide sequence complementary to any of the nucleotide sequences in (a).
  • nucleic acid molecules that comprise a polynucleotide having a nucleotide sequence at least 90% identical, and more preferably at least 95%, 96%, 97%, 98% or 99% identical to any of the nucleotide sequences in (a) or (b) above.
  • Nucleotide, polynucleotide, or nucleic acid sequences(s) are understood to mean, according to the present invention, either a double-stranded DNA, a single- stranded DNA, or products of transcription of the said DNAs (e.g., RNA molecules).
  • the nucleic acid, polynucleotide, or nucleotide sequences can be isolated, purified (or partially purified), by separation methods including, but not limited to, ion-exchange chromatography, molecular size exclusion chromatography, affinity chromatography, or by genetic engineering methods such as amplification, cloning or subcloning.
  • the polynucleotide sequences can also contain one or more polynucleotides encoding heterologous polypeptide sequences (e.g., tags that facilitate purification of the polypeptides of the invention (see, for example, U.S. Patent No. 6,342,362, hereby incorporated by reference in its entirety; Altendorf, et al. 1999- WWW, 2000 "Structure and Function of the F 0 Complex of the ATP Synthase from Escherichia CoIi," J. of Experimental Biology 203:19-28, G. B.; Baneyx 1999 Biotechnology 10:411-21; Eihauer, et al. 2001 J. Biochem. Biophys.
  • heterologous polypeptide sequences e.g., tags that facilitate purification of the polypeptides of the invention
  • vectors containing one or more of the polynucleotides provided such as vectors containing nucleotides encoding biologically active A ⁇ peptide fragments.
  • the vectors can be vaccine, replication, or amplification vectors.
  • the polynucleotides are operably associated with regulatory elements capable of causing the expression of the polynucleotide sequences.
  • Such vectors include, among others, chromosomal, episomal and virus-derived vectors, e.g., vectors derived from bacterial plasmids, from bacteriophage, from transposons, from yeast episomes, from insertion elements, from yeast chromosomal elements, from viruses such as baculoviruses, papova viruses, such as SV40, vaccinia viruses, adenoviruses, fowl pox viruses, pseudorabies viruses and retroviruses, and vectors derived from combinations of the aforementioned vector sources, such as those derived from plasmid and bacteriophage genetic elements (e.g., cosmids and phagemids).
  • vectors derived from bacterial plasmids, from bacteriophage, from transposons, from yeast episomes, from insertion elements, from yeast chromosomal elements, from viruses such as baculoviruses, papova viruses, such as SV40, vaccinia viruses,
  • vectors can also comprise elements necessary to provide for the expression and/or the secretion of a polypeptide, such as a fragment of the A ⁇ peptide, encoded by the nucleotide sequences provided in a given host cell.
  • the vector can contain one or more elements selected from the group consisting of a promoter, signals for initiation of translation, signals for termination of translation, and appropriate regions for regulation of transcription, hi certain embodiments, the vectors can be stably maintained in the host cell and can, optionally, contain signal sequences directing the secretion of translated protein. Other embodiments provide vectors that are not stable in transformed host cells. Vectors can integrate into the host genome or be autonomously-replicating vectors.
  • a vector comprises a promoter operably linked to a protein or peptide-encoding nucleic acid sequence, one or more origins of replication, and, optionally, one or more selectable markers (e.g., an antibiotic resistance gene).
  • selectable markers e.g., an antibiotic resistance gene.
  • Non-limiting exemplary vectors for the expression of polypeptides include pBr-type vectors, pET-type plasmid vectors (PROMEGA), pBAD plasmid vectors (INVITROGEN) or those provided in the examples below.
  • vectors are useful for transforming host cells for the cloning or expression of the nucleotide sequences provided.
  • Promoters which may be used to control expression include, but are not limited to, the CMV promoter, the SV40 early promoter region (Bernoist and Chambon 1981 Nature 290:304-310), the promoter contained in the 3' long terminal repeat of Rous sarcoma virus (Yamamoto, et al. 1980 Cell 22:787-797), the herpes thymidine kinase promoter (Wagner et al. 1981 Proc. Natl. Acad. Sci. USA 78:1441- 1445), the regulatory sequences of the metallothionein gene (Brinster et al.
  • prokaryotic vectors containing promoters such as the ⁇ -lactamase promoter (Villa-Kamaroff, et al. 1978 Proc. Natl. Acad. Sci. USA 75:3727-3731), or the tac promoter (DeBoer, et al. 1983 Proc. Natl. Acad. Sci. USA 80:21-25); see also, "Useful Proteins from Recombinant Bacteria" in Scientific American, 1980, 242:74- 94; plant expression vectors comprising the nopaline synthetase promoter region (Herrera-Estrella et al.
  • the cauliflower mosaic virus 35S RNA promoter (Gardner, et al. 1981 Nucl. Acids Res. 9:2871), and the promoter of the photosynthetic enzyme ribulose biphosphate carboxylase (Herrera-Estrella et al. 1984 Nature 310:115-120); promoter elements from yeast or fungi such as the Gal 4 promoter, the ADC (alcohol dehydrogenase) promoter, PGK (phosphoglycerol kinase) promoter, and/or the alkaline phosphatase promoter.
  • yeast or fungi such as the Gal 4 promoter, the ADC (alcohol dehydrogenase) promoter, PGK (phosphoglycerol kinase) promoter, and/or the alkaline phosphatase promoter.
  • nucleotide sequences are “homologous” or “modified” nucleotide sequences.
  • Modified nucleic acid sequences will be understood to mean any nucleotide sequence obtained by mutagenesis according to techniques well known to persons skilled in the art, and exhibiting modifications in relation to the normal sequences. For example, mutations in the regulatory and/or promoter sequences for the expression of a polypeptide that result in a modification of the level of expression of a polypeptide provide for a "modified nucleotide sequence”. Likewise, substitutions, deletions, or additions of nucleic acid to the polynucleotides provide for "homologous" or "modified” nucleotide sequences.
  • homologous or “modified” nucleic acid sequences have substantially the same biological or serological activity as the native (naturally occurring) A ⁇ peptide fragments.
  • a “homologous” or “modified” nucleotide sequence will also be understood to mean a splice variant of the polynucleotides of the instant invention or any nucleotide sequence encoding a "modified polypeptide" as defined below.
  • a homologous nucleotide sequence encompasses a nucleotide sequence having a percentage identity with the bases of the nucleotide sequences of between at least (or at least about) 80.0% to 99.9% (inclusive), or 85% to 99%, or 90% to 99%, or 95% to 99%.
  • homologous sequences exhibiting a percentage identity with the bases of the nucleotide sequences described can have 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99 percent identity with the polynucleotide sequences of the instant invention.
  • Both protein and nucleic acid sequence homologies may be evaluated using any of the variety of sequence comparison algorithms and programs known in the art. Such algorithms and programs include, but are by no means limited to, TBLASTN, BLASTP, FASTA, TFASTA, and CLUSTALW (Pearson and Lipman 1988 Proc. Natl. Acad. Sci. U.S.A. 85(8):2444-2448; Altschul, et al. 1990 J. MoI. Biol. 215(3):403-410; Thompson, et al. 1994 Nucleic Acids Res. 22(2):4673-4680; Higgins, et al. 1996 Methods Enzymol. 266:383-402; Altschul, et al. 1990 J. MoI. Biol. 215(3):403-410; Altschul, et al. 1993 Nature Genetics 3:266-272).
  • sequence comparison algorithms and programs include, but are by no means limited to, TBLASTN, BLASTP, FASTA, TFASTA,
  • nucleotide sequences complementary to any of the polynucleotide sequences disclosed herein are understood to include any DNA whose nucleotides are complementary to those of the sequence of the invention, and whose orientation is reversed (e.g., an antisense sequence).
  • fragments of the polynucleotide sequences disclosed herein are fragments of the polynucleotide sequences disclosed herein.
  • Representative fragments of the polynucleotide sequences will be understood to mean any nucleotide fragment having at least 8 or 9 successive nucleotides, preferably at least 12 successive nucleotides, and still more preferably at least 15 or at least 20 successive nucleotides of the sequence from which it is derived.
  • the upper limit for such fragments is the total number of polynucleotides found in the sequence encoding for A ⁇ i -42 peptide, (or, in certain embodiments, the open reading frame (ORF) identified herein).
  • the appropriate fragments thereof encoding for a specific peptide are also useful.
  • nucleotide sequences that are A ⁇ peptide fragment homologs, or fragments thereof, which have been previously identified can be utilized to carry out the method for inhibiting angiogenesis of the subject invention.
  • detection probes e.g., fragments of the disclosed polynucleotide sequences
  • Such a detection probe will advantageously have as sequence a sequence of at least 9, 12, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, or 100 nucleotides.
  • detection probes can comprise 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119,
  • the detection probes can also be used as labeled probe or primer in the subject invention.
  • Labeled probes or primers are labeled with a radioactive compound or with another type of label.
  • non-labeled nucleotide sequences may be used directly as probes or primers; however, the sequences are generally labeled with a radioactive element ( P, S, H, I) or with a molecule such as biotin, acetylaminofluorene, digoxigenin, 5-bromo- deoxyuridine, or fluorescein to provide probes that can be used in numerous applications.
  • nucleotide sequences disclosed may also be used in analytical systems, such as DNA chips.
  • DNA chips and their uses are well known in the art and (see for example, U.S. Patent Nos. 5,561,071; 5,753,439; 6,214,545; Schena, et al. 1996 BioEssays 18:427-431; Bianchi, et al. 1997 Clin. Diagn. Virol. 8:199-208; each of which is hereby incorporated by reference in their entireties) and/or are provided by commercial vendors such as AFFYMETRIX, Inc. (Santa Clara, Calif.).
  • hybridization is conducted under moderate to high stringency conditions by techniques well known in the art, as described, for example, in Keller, G. H., M. M. Manak 1987 DNA Probes, Stockton Press, New York, N. Y., pp. 169-170.
  • hybridization of immobilized DNA on Southern blots with 32 P-labeled gene-specific probes can be performed by standard methods (Maniatis, et al. 1982 Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York). In general, hybridization and subsequent washes can be carried out under moderate to high stringency conditions that allow for detection of target sequences with homology to the exemplified polynucleotide sequence. For double-stranded DNA gene probes, hybridization can be carried out overnight at 20- 25°C below the melting temperature (Tm) of the DNA hybrid in 6x SSPE, 5x Denhardt's solution, 0.1% SDS, 0.1 mg/ml denatured DNA. The melting temperature is described by the following formula (Beltz et al. 1983 Methods of Enzymology, R. Wu, L. Grossman and K. Moldave [eds.] Academic Press, New York 100:266-285).
  • T m 81.5°C.+16.6 Log[Na+]+0.41(%G+C)-0.61(% formamide)-600/length of duplex in base pairs. [0091] Washes are typically carried out as follows:
  • T m melting temperature
  • T m (°C) 2 (number T/A base pairs)+4 (number QIC base pairs) (Suggs et al. 1981 ICN- UCLA Symp. Dev. Biol. Using Purified Genes, D. D. Brown [ed.], Academic Press, New York, 23:683-693).
  • Washes can be carried out as follows:
  • salt and/or temperature can be altered to change stringency.
  • a labeled DNA fragment >70 or so bases in length the following conditions can be used:
  • procedures using conditions of high stringency can also be performed as follows: Pre-hybridization of filters containing DNA is carried out for 8 h to overnight at 65 0 C. in buffer composed of 6x SSC, 50 mM Tris-HCl (pH 7.5), 1 niM EDTA, 0.02% PVP, 0.02% Ficoll, 0.02% BSA, and 500 ⁇ g/ml denatured salmon sperm DNA. Filters are hybridized for 48 h at 65 0 C, the preferred hybridization temperature, in pre-hybridization mixture containing 100 ⁇ g/ml denatured salmon sperm DNA and 5-2OxIO 6 cpm of 32 P-labeled probe.
  • the hybridization step can be performed at 65 0 C. in the presence of SSC buffer, Ix SSC corresponding to 0.15M NaCl and 0.05 M Na citrate.
  • filter washes can be done at 37° C. for 1 h in a solution containing 2x SSC, 0.01% PVP, 0.01% Ficoll, and 0.01% BSA, followed by a wash in O.lx SSC at 50° C. for 45 min.
  • filter washes can be performed in a solution containing 2x SSC and 0.1% SDS, or 0.5x SSC and 0.1% SDS, or O.lx SSC and 0.1% SDS at 68° C. for 15 minute intervals.
  • the hybridized probes are detectable by autoradiography.
  • Other conditions of high stringency which may be used are well known in the art (see, for example, Sambrook, et al. 1989 Molecular Cloning, A Laboratory Manual, Second Edition, Cold Spring Harbor Press, N.Y., pp. 9.47-9.57; and Ausubel, et al. 1989 Current Protocols in Molecular Biology, Green Publishing Associates and Wiley Interscience, N.Y., each incorporated herein in its entirety).
  • a further non-limiting example of procedures using conditions of intermediate stringency are as follows: Filters containing DNA are pre-hybridized, and then hybridized at a temperature of 60° C. in the presence of a 5x SSC buffer and labeled probe. Subsequently, filters washes are performed in a solution containing 2x SSC at 50. degree. C. and the hybridized probes are detectable by autoradiography.
  • Other conditions of intermediate stringency which may be used are well known in the art (see, for example, Sambrook et al. 1989 Molecular Cloning, A Laboratory Manual, Second Edition, Cold Spring Harbor Press, N.Y., pp. 9.47-9.57; and Ausubel et al 1989 Current Protocols in Molecular Biology, Green Publishing Associates and Wiley Interscience, N. Y., each of which is incorporated herein in its entirety).
  • the probe sequences of the subject invention include mutations (both single and multiple), deletions, insertions of the described sequences, and combinations thereof, wherein said mutations, insertions and deletions permit formation of stable hybrids with the target polynucleotide of interest. Mutations, insertions and deletions can be produced in a given polynucleotide sequence in many ways, and these methods are known to an ordinarily skilled artisan. Other methods may become known in the future.
  • restriction enzymes can be used to obtain functional fragments of the subject DNA sequences.
  • BaBl exonuclease can be conveniently used for time-controlled limited digestion of DNA (commonly referred to as "erase-a-base” procedures). See, for example, Maniatis, et al. 1982 Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York; Wei, et al. 1983 J. Biol. Chem. 258:13006-13512.
  • the nucleic acid sequences disclosed can also be used as molecular weight markers in nucleic acid analysis procedures.
  • transformed cells comprise an expression vector containing polynucleotide sequences for an A ⁇ peptide fragment.
  • Other embodiments provide for host cells transformed with nucleic acids.
  • Yet other embodiments provide transformed cells comprising an expression vector containing fragments of A ⁇ polynucleotide sequences.
  • Transformed host cells can be cultured under conditions allowing the replication and/or the expression of the nucleotide sequences provided. Expressed polypeptides are recovered from culture media and purified, for further use, according to methods known in the art.
  • the host cell may be chosen from eukaryotic or prokaryotic systems, for example bacterial cells (Gram negative or Gram positive), yeast cells, animal cells, plant cells, and/or insect cells using baculovirus vectors.
  • the host cell for expression of the polypeptides include, and are not limited to, those taught in U.S. Patent Nos. 6,319,691; 6,277,375; 5,643,570; 5,565,335; Unger, et al. 1997 The Computer l l(17):20; or Smith, et al. 1998 The Engineer 12(22):20, each of which is incorporated by reference in its entirety, including all references cited within each respective patent or reference.
  • host cells include Staphylococcus spp., Enterococcus spp., E. coli, and Bacillus subtilis; fungal cells, such as Streptomyces spp., Aspergillus spp., S. cerevisiae, Schizosaccharomyces pombe, Pichia pastoris, Hansela polymorpha, Kluveromyces lactis, and Yarrowia lipolytica; insect cells such as Drosophila S2 and Spodoptera Sf9 cells; animal cells such as CHO, COS, HeLa, C127, 3T3, BHK, 293 and Bowes melanoma cells; and plant cells.
  • a great variety of expression systems can be used to produce the polypeptides provided and polynucleotides can be modified according to methods known in the art to provide optimal codon usage for expression in a particular expression system.
  • a host cell strain may be chosen that modulates the expression of the inserted sequences, modifies the gene product, and/or processes the gene product in the specific fashion. Expression from certain promoters can be elevated in tiie presence of certain inducers; thus, expression of the genetically engineered polypeptide may be controlled.
  • different host cells have characteristic and specific mechanisms for the translational and post-translational processing and modification (e.g., glycosylation, phosphorylation) of proteins. Appropriate cell lines or host systems can be chosen to ensure the desired modification and processing of the foreign protein expressed. For example, expression in a bacterial system can be used to produce an unglycosylated core protein product whereas expression in yeast will produce a glycosylated product. Expression in mammalian cells can be used to provide "native" glycosylation of a heterologous protein. Furthermore, different vector/host expression systems may effect processing reactions to different extents.
  • Nucleic acids and/or vectors can be introduced into host cells by well- known methods, such as, calcium phosphate transfection, DEAE-dextran mediated transfection, transfection, microinjection, cationic lipid-mediated transfection, electroporation, transduction, scrape loading, ballistic introduction and infection (see, for example, Sambrook, et al. 1989 Molecular Cloning: A Laboratory Manual, 2.sup.nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N. Y.).
  • the subject invention also provides for the expression of a polypeptide, derivative, or a variant (e.g., a splice variant) encoded by a polynucleotide sequence disclosed herein.
  • the invention provides for the expression of a polypeptide fragment obtained from a polypeptide, derivative, or a variant encoded by a polynucleotide fragment derived from the polynucleotide sequences disclosed herein.
  • the disclosed sequences can be regulated by a second nucleic acid sequence so that the polypeptide or fragment is expressed in a host transformed with a recombinant DNA molecule according to the subject invention.
  • the subject invention also provides nucleic acid-based methods for the identification of the presence of the A ⁇ gene, or fragments or variants thereof, in a sample. These methods can utilize the nucleic acids provided and are well known to those skilled in the art (see, for example, Sambrook, et al. 1989 Molecular Cloning, A Laboratory Manual, Second Edition, Cold Spring Harbor Press, N.Y., pp. 9.47-9.57, or Abbaszadega, et al. 2001 Reviews in Biology and Biotechnology, l(2):21-26).
  • nucleic acids can be used to screen individuals for disorders associated with dysregulation of the A ⁇ gene or its transcriptional products.
  • the subject invention also provides polypeptides encoded by nucleotide sequences of the invention.
  • the subject invention also provides fragments of at least 5 amino acids of a polypeptide encoded by the polynucleotides of the instant invention.
  • administering refers to the process of delivering an agent to a patient.
  • the process of administration can be varied, depending on the agent, or agents, and the desired effect.
  • Administration can be accomplished by any means appropriate for the therapeutic agent, for example, by oral, parenteral, mucosal, pulmonary, topical, catheter-based, rectal, intracranial, intracerebroventricular, intracerebral, intravaginal or intrauterine delivery.
  • Parenteral delivery can include for example, subcutaneous intravenous, intrauscular, intraarterial, and injection into the tissue of an organ, particularly tumor tissue.
  • Mucosal delivery can include, for example, intranasal delivery.
  • Oral or intranasal delivery can include the administration of a propellant.
  • Pulmonary delivery can include inhalation of the agent.
  • Catheter-based delivery can include delivery by iontropheretic catheter- based delivery.
  • Oral delivery can include delivery of a coated pill, or administration of a liquid by mouth.
  • Administration can generally also include delivery with a pharmaceutically acceptable carrier, such as, for example, a buffer, a polypeptide, a peptide, a polysaccharide conjugate, a liposome, and/or a lipid.
  • Gene therapy protocol is also considered an administration in which the therapeutic agent is a polynucleotide capable of accomplishing a therapeutic goal when expressed as a transcript or a polypeptide into the patient.
  • the A ⁇ peptide fragment is administered in an effective amount to inhibit pathological angiogenesis.
  • angiogenesis is intended to refer to the process by which new blood vessels are formed and which is essential to a variety of normal body activities (such as reproduction, development, and wound repair). The process is believed to involve a complex interplay of molecules which both stimulate and inhibit the growth of endothelial cells, the primary cells of the capillary blood vessels. Under normal conditions, these molecules appear to maintain the microvasculature in a quiescent state (i.e., one of no capillary growth) for prolonged periods. When necessary, however (such as during wound repair), these cells can undergo rapid proliferation and turnover within a short period of time.
  • angiogenesis is a highly regulated process under normal conditions, many conditions (characterized as “angiogenic diseases") are driven by persistent unregulated angiogenesis. Otherwise stated, unregulated angiogenesis may either cause a particular pathological condition directly or exacerbate an existing pathological condition. For example, ocular neovascularization has been implicated as the most common cause of blindness and dominates approximately twenty eye diseases. In certain existing conditions, such as arthritis, newly formed capillary blood vessels invade the joints and destroy cartilage. In diabetes, new capillaries formed in the retina invade the vitreous, bleed, and cause blindness.
  • tumors which enlarge to greater than 2 mm, must obtain then- own blood supply and do so by inducing the growth of new capillary blood vessels. Once these new blood vessels become embedded in the tumor, they provide a means for tumor cells to enter the circulation and metastasize to distant site, such as liver, lung or bone (Weidner, N. et al., The New England Journal of Medicine, 324(l):l-8, 1991).
  • compositions of the subject invention can be formulated according to known methods for preparing pharmaceutically useful compositions.
  • Formulations are described in a number of sources which are well known and readily available to those skilled in the art.
  • Remington's Pharmaceutical Sciences (Martin E W 1995 Easton Pennsylvania Mack Publishing Company, 19.sup.th ed.) describes formulations which can be used in connection with the subject invention.
  • Formulations suitable for parenteral administration include, for example, aqueous sterile injection solutions, which may contain antioxidants, buffers, bacteriostats, and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and nonaqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations maybe presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze dried (lyophilized) condition requiring only the condition of the sterile liquid carrier, for example, water for injections, prior to use.
  • sterile liquid carrier for example, water for injections, prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powder, granules, tablets, etc.
  • the formulations of the subject invention can include other agents conventional in the art having regard to the type of formulation in question.
  • the A ⁇ peptide fragments are delivered in a sustained release formulation.
  • the formulations provide extended release and extended half- life.
  • Controlled release systems suitable for use include, without limitation, diffusion- controlled, solvent-controlled and chemically-controlled systems.
  • Diffusion controlled systems include, for example reservoir devices, in which the A ⁇ peptide fragment or fragments are enclosed within a device such that release of the peptide fragments is controlled by permeation through a difussion barrier.
  • Common reservoir devices include, for example, membranes, capsules, microcapsules, liposomes, and hollow fibers.
  • Monolithic (matrix) device are a second type of diffusion controlled system, wherein the A ⁇ peptide fragment(s) are dispersed or dissolved in an rate- controlling matrix (e.g., a polymer matrix).
  • rate- controlling matrix e.g., a polymer matrix
  • the peptide fragments are homogeneously dispersed throughout a rate-controlling matrix and the rate of drug release is controlled by diffusion through the matrix.
  • Polymers suitable for use in the monolithic matrix device include naturally occurring polymers, synthetic polymers and synthetically modified natural polymers, as well as polymer derivatives.
  • Therapeutically effective and optimal dosage ranges for the A ⁇ peptide fragments can be determined using methods known in the art. Guidance as to appropriate dosages to achieve an anti-angiogenesis and/or anti-tumor effect is provided from the exemplified assays disclosed herein. The minimal amounts of A ⁇ peptide fragment to achieve a therapeutic effect can likewise be determined.
  • the A ⁇ peptide fragment is administered in an equivalent amount to be within the ⁇ M dose range. In another embodiment, an amount equivalent to about 1 ⁇ M to about 100 ⁇ M A ⁇ peptide fragment is administered. In another embodiment, an amount equivalent to about 2 ⁇ M to about 10 ⁇ M A ⁇ peptide fragment is administered.
  • Pharmaceutical formulations that can be administered can comprise, e.g., 1-10,000 mg, 10-1000 mg, 50-900 mg, 100-800 mg, or 200-500 mg.
  • the subject invention also pertains to diagnostic and/or screening methods and kits to screen for compounds that are potentially therapeutic in treatment of Alzheimer's disease by interfering with the anti-angiogenic effect of an A ⁇ peptide fragment.
  • a method for identifying compounds that interfere with A ⁇ -induced angiogenesis inhibition includes the steps of (a) contacting a first biological sample capable of undergoing angiogenesis with a test compound, a biologically active amount of an A ⁇ peptide fragment, and an angiogenic agent; and (b) determining the extent of angiogenesis that occurs in the first biological sample.
  • the method can include the steps of: (c) separately contacting a second biological sample capable of undergoing angiogenesis with a biologically active amount of an A ⁇ peptide fragment and an angiogenic agent; (d) determining the extent of angiogenesis that occurs in the second biological sample; and (e) comparing the extent of angiogenesis that occurs in the first biological sample with that which occurs in the second biological sample.
  • steps (c)-(d) can be utilized as a control.
  • the same A ⁇ peptide fragment is used in the first and second biological samples.
  • Determining the extent of angiogenesis can be carried out using methods known in the art, such as those described herein, and can be done qualitatively or quantitatively. For example, molecular or cellular markers of cancer or tumor growth can be utilized. The extent of angiogenesis can also be determined by measuring the amount of endothelial cell proliferation or the extent of blood vessel growth within a biological sample.
  • the biological samples utilized in the methods and kits can include various biological fluids and tissues that can exhibit angiogenesis and/or tumor development.
  • the biological sample can be arterial tissue, corneal tissue, endothelial cells, umbilical cord tissue, chorionic allantoid membrane, and the like.
  • the angiogenic agent can be any molecule, compound, or cell that is capable of inducing angiogenesis in the biological sample.
  • the angiogenic agent can be a trophic factor, such as a neurotrophic factor.
  • the angiogenic factor can be a cytokine or growth factor such as vascular endothelial growth factor, platelet-derived growth factor, and basic fibroblast growth factor.
  • the diagnostic and/or screening methods of the subject invention can be carried out in vivo, such as in an animal model, or in vitro.
  • the subject invention includes a kit for identifying compounds that interfere with A ⁇ -induced angiogenesis inhibition.
  • the kit can include a compartment containing at least one A ⁇ peptide fragment and, optionally, a compartment containing an angiogenic agent.
  • the kit can optionally include a compartment containing one or more biological samples.
  • a method for identifying compounds that interfere with A ⁇ -induced anti-tumor activity including the steps of: (a) contacting a first tumor tissue with a test compound and a biologically active amount of an A ⁇ peptide fragment; and (b) determining the extent of tumor growth that occurs in the tumor tissue.
  • the method can further include the steps of: (c) separately contacting a second tumor tissue with a biologically active amount of an A ⁇ peptide fragment; (d) determining the extent of tumor growth that occurs in the second tumor tissue; and (e) comparing the extent of tumor growth that occurs in the first tumor tissue with that which occurs in the second tumor tissue, hi this way, steps (c)-(d) can be utilized as a control.
  • the extent of tumor growth can be determined quantitatively or qualitatively using methods known in the art, including methods described herein. For example, molecular or cellular markers of cancer or tumor growth can be utilized.
  • the subject invention includes a kit for identifying compounds that interfere with A ⁇ -induced anti-tumor activity.
  • the kit can include a compartment containing at least one A ⁇ peptide fragment and, optionally, a compartment containing at least one tumor tissue. Furthermore, the kit can optionally include a compartment containing one or more biological samples.
  • the test compounds that can be screened using the methods and kits of the subject invention can include any substance, agent, or molecule, including, for example, small molecules and living or dead cells.
  • a variety of patients may be treated including any vertebrate species.
  • the patient is of a mammalian species.
  • Mammalian species which benefit from the disclosed methods of treatment include, and are not limited to, apes, chimpanzees, orangutans, humans, monkeys; domesticated animals (e.g., pets) such as dogs, cats, guinea pigs, hamsters, Vietnamese pot-bellied pigs, rabbits, and ferrets; domesticated farm animals such as cows, buffalo, bison, horses, donkey, swine, sheep, and goats; exotic animals typically found in zoos, such as bear, lions, tigers, panthers, elephants, hippopotamus, rhinoceros, giraffes, antelopes, sloth, gazelles, zebras, wildebeests, prairie dogs, koala bears, kangaroo, opossums, raccoons, pandas
  • a method for treating tumors, cancers or other proliferative disorders in animals or humans in need of such treatment comprising administering a therapeutically effective amount, optionally in unit dosage form, of an A ⁇ peptide fragment described herein.
  • methods for inhibiting angiogenesis in animals or humans in need thereof comprising administering a therapeutically effective amount, optionally in unit dosage form, of a an A ⁇ peptide fragment disclosed herein.
  • a ⁇ peptide fragments and pharmaceutical compositions comprising the fragments are provided, that can be used in one embodiment to treat tumors and cancers, including, but not limited to cancers or tumors in the following tissues or organs: breast, prostate, lung, bronchus, colon, urinary tract, bladder, kidney, pancreas, thyroid, stomach, brain, esophagus, liver, intrahepatic bile duct, cervix, skin, larynx, heart, testis, small intestine, thyroid, vulva, gallbladder, pleura, eye, nose, ear, nasopharnx, ureter, gastrointestineal system, rectal tissue, pancreas, head and neck.
  • Cancers that can be treated include without limitation non-Hodgkin lymphoma, melanoma, multiple myeloma, acute myeloid leukemia, chronic lymphatic leukemia, Hodgkin lymphoma, chronic myeloid leukemia, acute lymphatic leukemia, carcinomas, adenocarcinomas; sarcomas; lymphomas, and leukemias.
  • the A ⁇ peptide fragments can be used to treat, for example, prostate cancer, lung cancer, colorectal cancer, bladder cancer, cutaneous melanoma, pancreatic cancer, leukemia, breast cancer, endometrial cancer, non- Hodgkin's lymphoma, and ovarian cancer.
  • the A ⁇ peptide fragments can be used to treat epithelial cell cancers and tumors including: skin cancer, cervical cancer, anal carcinoma, esophageal cancer, hepatocellular carcinoma (in the liver), laryngeal cancer, renal cell carcinoma (in the kidneys), stomach cancer, testicular cancers, and thyroid cancer.
  • the A ⁇ peptide fragments are used to treat hematological malignancies (blood and bone marrow) including leukemia, lymphoma, and multiple myeloma.
  • the A ⁇ peptide fragments are used to treat sarcomas including: osteosarcoma (in bone), chondrosarcoma (arising from cartilage), and rhabdomyosarcoma (in muscle).
  • the A ⁇ peptide fragments are used to treat cancers and tumors of miscellaneous origin including: brain tumors, gastrointestinal stromal tumors (GIST), mesothelioma (in the pleura or pericardium), thymoma and teratomas, and melanoma.
  • GIST gastrointestinal stromal tumors
  • mesothelioma in the pleura or pericardium
  • thymoma and teratomas and melanoma.
  • tumors examples include, without limitation, malignant brain tumors, such as glioblastomas; malignant lung tumors, such as adenocarcinomas; or malignant tumors of the breast, colon, kidney, bladder, head or neck.
  • malignant brain tumors such as glioblastomas
  • malignant lung tumors such as adenocarcinomas
  • malignant tumors of the breast, colon, kidney, bladder, head or neck include, without limitation, malignant brain tumors, such as glioblastomas; malignant lung tumors, such as adenocarcinomas; or malignant tumors of the breast, colon, kidney, bladder, head or neck.
  • Proliferative disorders that can be treated include, without limitation, hematopoietic disorders, such as leukemias, lymphomas or polycythemias; and ocular disorders, such as diabetic retinopathy, macular degeneration, glaucoma or retinitis pigmentosa.
  • Inflammatory disorders that can be treated include, without limitation, rheumatoid arthritis, osteoarthritis, pulmonary fibrosis, sarcoid granulomas, psoriasis or asthma.
  • the A ⁇ peptide fragments can be used to treat a carcinoma, sarcoma, lymphoma, leukemia, and/or myeloma. In other embodiments, the A ⁇ peptide fragments disclosed herein can be used to treat solid tumors.
  • the A ⁇ peptide fragments described herein can be used for the treatment of cancer, including, but not limited to the cancers listed in Table 2a below.
  • HHQKLVFF is the sequence of A ⁇ that confers anti-antiogenic activity.
  • Numerous studies have shown that heparin and various proteoglycans on the cell surface can bind to A ⁇ peptides (Snow, et al. 1995 Arch. Biochem. Biophys. 320, 84-95; McLaurin, et al. 2000 Eur. J. Biochem. 267, 6353-61; McKeon J, Holland LA. 2004 Electrophoresis 25, 1243-8), and heparan sulfate proteoglycans have been shown to be associated with amyloid deposits in AD brain (van Horssen, et al.
  • Heparin sulfate proteoglycans also play a prominent role during angio genesis by allowing the interaction of specific growth factors such as basic fibroblast growth factor (bFGF) and vascular endothelial growth factor (VEGF) with the cell surface. In this way, proteoglycans are thought to modulate the interaction of growth factors with receptors (Rusnati M, Presta M. 1996 Int. J. Clin. Lab. Res. 26, 15-23; Dougher, et al. 1997 Growth Factors. 14, 257-68).
  • bFGF basic fibroblast growth factor
  • VEGF vascular endothelial growth factor
  • Cell surface proteoglycans such as heparan sulfate proteoglycans can bind to and allow the activity of various growth factors including VEGF and bFGF (Iozzo RV, San Antonio JD. 2001 J. Clin. Invest. 108, 349-55; Presta, et al. 2005 Cytokine Growth Factor Rev. 16, 159-78; Sanderson, et al. 2005 J. Cell Biochem. Sep 7, (advance electronic publication)). It is possible that A ⁇ binds to these proteoglycans, impacting the binding and interaction of growth factors with the cell.
  • VEGF vascular endothelial growth factor
  • angiogenesis assays contain heparin binding growth factors
  • the addition of excess heparin may act to bind out A ⁇ peptides and prevent their binding to the cell surface, hence opposing the anti-angiogenic activity of A ⁇ .
  • a ⁇ has also been shown to directly interact with the heparin binding motif on VEGF (Yang, et al. 2005 J. Neurochem. 93, 118-27); hence it is possible that the binding of A ⁇ to heparin can prevent it from binding to VEGF, reversing the anti-angiogenic activity of A ⁇ .
  • heparin and other glycosaminoglycans affect the conformational properties of A ⁇ peptides, changing the rate of fibril formation (Castillo, et al. 1999 J. Neurochem. 72, 1681-7; Cohlberg, et al. 2002 Biochemistry. 41, 1502-11) thereby rendering the peptide unable to block angiogenesis.
  • the anti-angiogenic activity of A ⁇ peptides in-vitro seems to be related to their conformational properties, as preparations of A ⁇ containing higher ⁇ -sheet content are more potently anti- angiogenic (Gebbink, et al. 2000 Biochim. Biophys. Acta. 1502, 16-30.
  • soluble oligomers of the peptide are particularly anti-angiogenic whereas fibrillar forms are inactive (Paris, et al. 2005 Brain Res. MoI. Brain Res. 136, 212-30; Skovseth, et al. 2005 Blood 105, 1044-51) suggesting that particular residues in the A ⁇ peptide need to be exposed in order to inhibit angiogenesis.
  • HHQK putative proteoglycan binding region
  • HHQK Carlson, A.D.; Weintaub, H.J.R. 1989 Arteriosclerosis. 9, 21-32; Snow, et al.1995 Arch. Biochem. Biophys. 320, 84-95; McLaurin , et al. 2000 Eur. J. Biochem. 267, 6353-61; McKeon, et al. 2004 Electrophoresis. 25, 1243-8).
  • Proteoglycans are known to play a regulatory role during angiogenesis (Moon, et al. 2005 J. Cell Physiol.
  • VEGF is neurotrophic ⁇ it is important for maintaining vascular integrity, and also a key factor in vascular remodeling following stroke or head injury (Slevin, et al. 2000 Neuroreport 11, 2759-64; Shore, et al. 2004 Neurosurgery. 54, 605-12).
  • AD patients and transgenic mouse models of AD do poorly following stroke (Koistinaho, et al. 2002 Proc. Natl. Acad. Sci. U.S.A. 99, 1610-5; Wen, et al. 2004 J. Biol. Chem. 279, 22684-92; Koistinaho M, Koistinaho J. 2005 Brain Res. Brain Res. Rev. 48, 240-50).
  • Examples provided herein support that the proteoglycan binding motif alone may not be sufficient to elicit anti-angiogenic effects, and that the amino acids immediately adjacent to this sequence (LVFF) are required to mediate the anti- angiogenic activity of A ⁇ .
  • LVFF amino acids immediately adjacent to this sequence
  • the pro-angiogenic affects of A ⁇ 3 4-42 have also been noted.
  • the pro-angiogenic activity of the A ⁇ 34-42 fragment observed in the network assay described herein is consistent with the pro-angiogenic activity of A ⁇ i..io / 42 peptides at low concentrations that has previously observed (Paris, et al. 2004 Angiogenesis. 7, 75-85; Cantara, et al. 2004 F.A.S.E.B. J. 18, 1943-5).
  • the folding of A ⁇ may be such that the C-terminal 34-42 sequence is left exposed when monomers and dimers are formed. Subsequently this region may be buried upon higher order oligomer or fibril formation.
  • the peptide fragments disclosed herein can be used in combination with at least one additional chemotherapeutic agent in order to treat a cancer, tumor or other proliferative disorder.
  • the additional agents can be administered in combination or alternation with the compounds disclosed herein.
  • the drugs can form part of the same composition, or be provided as a separate composition for administration at the same time or a different time.
  • second therapeutic agents include but are not limited to, EL- 12, retinoids, interferons, angiostatin, endostatin, thalidomide, thrombospondin-1, thrombospondin-2, captopryl, anti-neoplastic agents such as alpha interferon, COMP (cyclophosphamide, vincristine, methotrexate and prednisone), etoposide, mBACOD (methortrexate, bleomycin, doxorubicin, cyclophosphamide, vincristine and dexamethasone), PRO-MACE/MOPP (prednisone, methotrexate (w/leucovin rescue), doxorubicin, cyclophosphamide, taxol, etoposide/mechlorethamine, vincristine, prednisone and procarbazine), vincristine, vinblastine, angioinhibins, TNP-
  • agents with antimitotic effects include agents with antimitotic effects (antimitotic inhibitors), such as those which target cytoskeletal elements, including microtubule modulators such as taxane drugs (such as taxol, paclitaxel, taxotere, docetaxel), podophylotoxins or vinca alkaloids (vincristine, vinblastine); antimetabolite drugs (such as 5-fluorouracil, cytarabine, gemcitabine, purine analogues such as pentostatin, methotrexate); alkylating agents or nitrogen mustards (such as nitrosoureas, cyclophosphamide or ifosphamide); drugs which target DNA such as the antracycline drugs adriamycin, doxorubicin, pharmorubicin or epirubicin; drugs which target topoisomerases (topoisomerase inhibitors) such as etoposide; hormones and hormone agonists or antagonists such as estrogens, antiestrog
  • Angiogenesis assays known in the art may be used. See, for example, U.S. Patent Application 2003/0077261A1 to Paris, et al. wherein rat aortic ring, bovine, mouse and human angiogenesis assays are described.
  • Endothelial Cell Migration Assays described in U.S. Patent Application 2003/007726 IAl to Paris, et al. may be used, where migration of human brain adult endothelial cells is evaluated using a modified Boyden chamber assay (BD BioCoat MATRIGEL Invasion Chamber), as described (Soker et al. 1998; Nakamura et al. 1997).
  • BD BioCoat MATRIGEL Invasion Chamber BD BioCoat MATRIGEL Invasion Chamber
  • Nude Mouse Tumor Xenograft models as described, for example, in U.S. Patent Application 2003/007726 IAl to Paris, et al. may be used wherein A-549 (human lung adenocarcinoma) and U87-MG (human glioblastoma) cells are implanted into 8-week-old female nude mice. Tumors grown in the animals are measuring before, after and during treatment with A ⁇ peptides. On the termination day of each in vivo antitumor study, tumors are extracted and microvessels are quantified.
  • Example 1 Cell Culture and Reagents
  • AU in-vitro experiments were performed using primary Human Umbilical Vein Endothelial Cells (HUVEC) at passages 3 - 4, purchased from American Tissue Type Culture Collection (ATCC, VA). Cells were cultured in F12K Medium (ATCC, VA) supplemented with 10% fetal bovine serum (Invitrogen, CA), 0.1 mg/ml Heparin and 0.03 mg/ml endothelial cell growth supplement (Sigma-Aldrich, MO). At all times, cells were maintained in a sterile cell culture incubator at 37°C and 5% CO 2 .
  • HUVEC Human Umbilical Vein Endothelial Cells
  • peptides were prepared by and purchased from Biosource, CA upon request. 1 mg of lyophilized peptides were dissolved in 1 ml of 1,1,1,3,3,3- hexafluoro-2-propanol (HFIP) in order to minimize formation of ⁇ -sheet structures and promote ⁇ -helical secondary structure. Peptides were allowed to air dry in a chemical fume hood for one hour, followed by further drying in a speed-vac (Thermo- Savant, NY) for 30 minutes. The resulting clear film was re-suspended in 100% dimethylsulfoxide (DMSO) to a concentration of ImM. Peptides were subsequently aliquoted and stored at -80 0 C.
  • DMSO dimethylsulfoxide
  • HUVEC 7.5 x 10 4 cells/ml in 500 ⁇ l of medium were seeded in 24-well plates, on top of a layer of Matrigel basement membrane matrix (Invitrogen, CA) in F12K medium (ATCC, VA) containing 4% serum (Invitrogen, CA), 0.1 mg/ml Heparin and 0.03 mg/ml endothelial cell growth supplement (Sigma-Aldrich, MO). Cells were incubated with peptides (or control conditions) for 24 hours. Control wells received the same volume of vehicle (DMSO) used to dilute the peptides. Network formation experiments were performed in triplicate, and at least 2 randomly chosen fields were photographed for each well using a 4X objective. Capillary length was measured using Image Pro Plus software (Media Cybernetic, Inc., MD).
  • Example 4 Cell Proliferation Assay
  • HUVEC (5 x 10 3 cells per well) were seeded in a 96 well plate. Cells were incubated with peptides (or control conditions) for 24 hours. A quick cell proliferation assay was performed as per the manufacturer's protocol (Biovision Inc., CA).
  • HUVEC (1 x 10 4 cells per well) were seeded in a 96 well plate pre-coated with basement membrane protein complex. Cells were incubated with peptides (or control conditions) for 2.5 hours. For measurement of cell adhesion, the Innocyte cell adhesion assay was used (Calbiochem, CA) and the protocol followed as per the manufacturer's recommendations.
  • Example 8 Effect of A ⁇ peptide fragments on capillary tube formation
  • a ⁇ peptides and peptide fragments were tested for their ability to inhibit capillary network formation in the assay described in Example 3, including A ⁇ 1-42 , A ⁇ 1-40 , A ⁇ i -28 , A ⁇ 12-28 , A ⁇ 17-28 , A ⁇ 25-35 , A ⁇ io- 35 , A ⁇ io-i6 and A ⁇ 34-42 ) at 1, 5 and lO ⁇ M.
  • Total length of capillary tubes was quantified for each treatment group (n > 8), and expressed as a percentage of control treatment ( Figure 1).
  • Example 9 Effect of A ⁇ peptide fragments on cell proliferation and cell adhesion
  • Example 11 Effect of proteoglycan binding region mutant A ⁇ peptide fragments on capillary tube formation
  • Example 12 Effect of LVFF mutant A ⁇ peptide fragments on capillary tube formation
  • VFF amino acid sequence adjacent on the C-terminal side of the HHQK sequence was established by testing peptide fragments consisting of 9 amino acids starting at the HHQK sequence (table 2, fragments 1-3) in the capillary tube formation assay described in Example 3.
  • Example 13 Effect of A ⁇ n- 28 peptide fragments in the rat corneal micropocket assay
  • Example 14 Effect of A ⁇ i2-28, A ⁇ -28 mutants and A ⁇ i3_2o in the rat corneal micropocket assay

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Neurology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Neurosurgery (AREA)
  • Psychiatry (AREA)
  • Cardiology (AREA)
  • Hospice & Palliative Care (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

La présente invention porte sur des fragments de peptide Aß qui sont utiles pour inhiber l'angiogenèse. Cette invention concerne également des méthodes de traitement de l'angiogenèse pathologique ou indésirable ainsi que des pathologies et des maladies associées à cette dernière, lesdites méthodes consistant à administrer une quantité efficace d'un fragment Aß. Dans une forme de réalisation particulière, le fragment de peptide contient la séquence HHQKLVFF.
PCT/US2006/043921 2005-11-10 2006-11-13 Modulation de l'angiogenese au moyen de fragments de peptide a-beta WO2007059000A2 (fr)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP06837407A EP1951750A4 (fr) 2005-11-10 2006-11-13 Modulation de l'angiogenèse au moyen de fragments de peptide a-beta
CA002619455A CA2619455A1 (fr) 2005-11-10 2006-11-13 Modulation de l'angiogenese au moyen de fragments de peptide a-beta
AU2006315686A AU2006315686A1 (en) 2005-11-10 2006-11-13 Modulation of angiogenesis by A-beta peptide fragments
JP2008540237A JP2009516654A (ja) 2005-11-10 2006-11-13 A−ベータペプチドによる血管新生の調節

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US73547205P 2005-11-10 2005-11-10
US60/735,472 2005-11-10

Publications (2)

Publication Number Publication Date
WO2007059000A2 true WO2007059000A2 (fr) 2007-05-24
WO2007059000A3 WO2007059000A3 (fr) 2009-05-14

Family

ID=38049186

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/043921 WO2007059000A2 (fr) 2005-11-10 2006-11-13 Modulation de l'angiogenese au moyen de fragments de peptide a-beta

Country Status (8)

Country Link
US (1) US8067372B2 (fr)
EP (1) EP1951750A4 (fr)
JP (1) JP2009516654A (fr)
KR (1) KR20080095836A (fr)
CN (1) CN101558080A (fr)
AU (1) AU2006315686A1 (fr)
CA (1) CA2619455A1 (fr)
WO (1) WO2007059000A2 (fr)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009143489A2 (fr) * 2008-05-22 2009-11-26 Archer Pharmaceuticals, Inc. Modulation d’angiogenèse par des fragments de peptide a-bêta
US8067372B2 (en) 2005-11-10 2011-11-29 Alzheimer's Institute Of America, Inc. Modulation of angiogenesis by A-beta peptide fragments
CN108350052A (zh) * 2015-11-09 2018-07-31 英属哥伦比亚大学 淀粉样蛋白β中间区域中的表位及其构象选择性抗体
US10751382B2 (en) 2016-11-09 2020-08-25 The University Of British Columbia Anti-amyloid beta antibodies binding to a cyclic amyloid beta peptide
US10759837B2 (en) 2015-11-09 2020-09-01 The University Of British Columbia Anti-amyloid beta antibodies binding to a cyclic amyloid beta peptide
US10772969B2 (en) 2015-11-09 2020-09-15 The University Of British Columbia N-terminal epitopes in amyloid beta and conformationally-selective antibodies thereto

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI239847B (en) * 1997-12-02 2005-09-21 Elan Pharm Inc N-terminal fragment of Abeta peptide and an adjuvant for preventing and treating amyloidogenic disease
US20080050367A1 (en) * 1998-04-07 2008-02-28 Guriq Basi Humanized antibodies that recognize beta amyloid peptide
MY139983A (en) * 2002-03-12 2009-11-30 Janssen Alzheimer Immunotherap Humanized antibodies that recognize beta amyloid peptide
CA2513722A1 (fr) * 2003-02-01 2004-08-19 Neuralab Limited Immunisation active permettant de produire des anticorps contre a-beta soluble
WO2006066089A1 (fr) 2004-12-15 2006-06-22 Neuralab Limited Anticorps du peptide beta-amyloide humanises utilises dans l'amelioration de la cognition
US8784810B2 (en) * 2006-04-18 2014-07-22 Janssen Alzheimer Immunotherapy Treatment of amyloidogenic diseases
WO2009017467A1 (fr) 2007-07-27 2009-02-05 Elan Pharma International Limited Traitement de maladies amyloïdogéniques
US8003097B2 (en) * 2007-04-18 2011-08-23 Janssen Alzheimer Immunotherapy Treatment of cerebral amyloid angiopathy
RU2523894C2 (ru) * 2007-04-18 2014-07-27 Янссен Альцгеймер Иммунотерапи Предупреждение и лечение церебральной амилоидной ангиопатии
EP2044951A1 (fr) * 2007-10-02 2009-04-08 Merz Pharma GmbH & Co. KGaA Utilisation de substances pour le traitement de la perte de la vue chez les humains avec un glaucome et autres maladies de l'oeil dégénératives
JO3076B1 (ar) * 2007-10-17 2017-03-15 Janssen Alzheimer Immunotherap نظم العلاج المناعي المعتمد على حالة apoe
US9067981B1 (en) 2008-10-30 2015-06-30 Janssen Sciences Ireland Uc Hybrid amyloid-beta antibodies
US10882890B2 (en) * 2014-07-25 2021-01-05 Academia Sinica Bipartite molecules and uses thereof in treating diseases associated with abnormal protein aggregates
IT202100023357A1 (it) 2021-09-09 2023-03-09 Cheirontech S R L Peptidi con attività anti-angiogenica

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3105009A (en) * 1959-10-30 1963-09-24 Us Rubber Co Anti-mitotic chemotherapeutic compounds
US5780587A (en) * 1990-08-24 1998-07-14 President And Fellows Of Harvard College Compounds and methods for inhibiting β-protein filament formation and neurotoxicity
US5434050A (en) 1991-08-13 1995-07-18 Regents Of The University Of Minnesota Labelled β-amyloid peptide and methods of screening for Alzheimer's disease
EP1298436B1 (fr) 1992-10-26 2010-07-14 Elan Pharmaceuticals, Inc. Procédés de l' identification des composés inhibiteurs de la libération du peptide beta-amyloide (BAP)
WO1994012627A1 (fr) 1992-11-25 1994-06-09 Cephalon, Inc. Modeles d'animaux transgeniques utilises dans le traitement de la maladie d'alzheimer
EP0815134B1 (fr) 1995-03-14 2002-06-05 Praecis Pharmaceuticals Incorporated Modulateurs de l'agregation de substances amyloides
EP0866805A1 (fr) 1995-12-12 1998-09-30 Karolinska Innovations AB PEPTIDE FIXANT LA SEQUENCE KLVFF DE L'AMYLOIDE $g(b)
US6043283A (en) * 1996-09-20 2000-03-28 Baylor College Of Medicine Tyramine compounds and their neuronal effects
US5981471A (en) 1997-02-06 1999-11-09 Entremed, Inc. Compositions and methods for inhibiting cellular proliferation
CN1433321A (zh) 1999-11-29 2003-07-30 尼奥切姆公司 预防和治疗Alzheimer症和其他与淀粉样蛋白有关的疾病的疫苗
US6998137B2 (en) * 2000-04-07 2006-02-14 Macromed, Inc. Proteins deposited onto sparingly soluble biocompatible particles for controlled protein release into a biological environment from a polymer matrix
EP1424892A4 (fr) * 2001-08-10 2006-08-23 Univ South Florida Modulation de l'angiogenese par des peptides a-beta
GB0207362D0 (en) 2002-03-28 2002-05-08 Univ Liverpool Chemotherapy
AU2006315686A1 (en) 2005-11-10 2007-05-24 Roskamp Research, Llc Modulation of angiogenesis by A-beta peptide fragments

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP1951750A4 *

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8067372B2 (en) 2005-11-10 2011-11-29 Alzheimer's Institute Of America, Inc. Modulation of angiogenesis by A-beta peptide fragments
WO2009143489A2 (fr) * 2008-05-22 2009-11-26 Archer Pharmaceuticals, Inc. Modulation d’angiogenèse par des fragments de peptide a-bêta
WO2009143489A3 (fr) * 2008-05-22 2010-03-11 Archer Pharmaceuticals, Inc. Modulation d’angiogenèse par des fragments de peptide a-bêta
CN108350052A (zh) * 2015-11-09 2018-07-31 英属哥伦比亚大学 淀粉样蛋白β中间区域中的表位及其构象选择性抗体
US10759837B2 (en) 2015-11-09 2020-09-01 The University Of British Columbia Anti-amyloid beta antibodies binding to a cyclic amyloid beta peptide
US10772969B2 (en) 2015-11-09 2020-09-15 The University Of British Columbia N-terminal epitopes in amyloid beta and conformationally-selective antibodies thereto
US10774120B2 (en) 2015-11-09 2020-09-15 The University Of British Columbia Anti-amyloid beta antibodies binding to a cyclic amyloid beta peptide
US11905318B2 (en) 2015-11-09 2024-02-20 The University Of British Columbia Cyclic compound/peptide comprising an A-beta13-16 peptide and a linker that is covalently coupled to the n-terminus residue and the c-terminus residue of the A-beta13-16 peptide
US11970522B2 (en) 2015-11-09 2024-04-30 The University Of British Columbia Cyclic compound/peptide comprising an A-beta15-18 peptide and a linker that is covalently coupled to the n-terminus residue and the c-terminus residue of the A-BETA15-18 peptide
US10751382B2 (en) 2016-11-09 2020-08-25 The University Of British Columbia Anti-amyloid beta antibodies binding to a cyclic amyloid beta peptide
US11779629B2 (en) 2016-11-09 2023-10-10 The University Of British Columbia Compositions comprising cyclic peptides derived from an A-beta peptide

Also Published As

Publication number Publication date
JP2009516654A (ja) 2009-04-23
CN101558080A (zh) 2009-10-14
EP1951750A4 (fr) 2009-12-09
WO2007059000A3 (fr) 2009-05-14
US20080031954A1 (en) 2008-02-07
AU2006315686A1 (en) 2007-05-24
KR20080095836A (ko) 2008-10-29
CA2619455A1 (fr) 2007-05-24
EP1951750A2 (fr) 2008-08-06
US8067372B2 (en) 2011-11-29

Similar Documents

Publication Publication Date Title
WO2007059000A2 (fr) Modulation de l'angiogenese au moyen de fragments de peptide a-beta
US20060147945A1 (en) Novel secreted proteins and their uses
US20050131212A1 (en) Neural regeneration peptides and methods for their use in treatment of brain damage
US20100266548A1 (en) LPS-Responsive CHS1/Beige-Like Anchor Gene and Therapeutic Applications Thereof
EP1309614A2 (fr) Nouvelles proteines secretees et leurs utilisations
US20040038242A1 (en) Novel secreted proteins and their uses
US20110306557A1 (en) Neural regeneration peptides and methods for their use in treatment of brain damage
US20020115610A1 (en) Prokineticin polypeptides, related compositions and methods
KR20120024741A (ko) 신경변성 질환과 알츠하이머병을 치료하고 정상 기억을 향상시키기 위한 방법과 조성물
JP4969006B2 (ja) 神経細胞死を抑制するポリペプチド、Humanin
US7589168B2 (en) Inhibition of angiogenesis by A-β peptides
US6617430B2 (en) Ataxin-2 binding proteins
US20110207911A1 (en) Polypeptide derived from the ryanodine receptor that inhibits calcium leakage
WO2009143489A2 (fr) Modulation d’angiogenèse par des fragments de peptide a-bêta
JP2008013436A (ja) 血管形成促進剤
US20100130416A1 (en) Modulation of angiogenesis by a-beta peptide fragments
JP4707235B2 (ja) Tiam2(t細胞のリンパ腫の侵襲およびmetastasis2)ヌクレオチド交換因子
KR100508845B1 (ko) KVLQT1을 코딩하는 장기 QT증후군 유전자 및 KVLQT1과 minK의 결합체
US20040215007A1 (en) Novel secreted proteins and their uses
EP1529843A2 (fr) Protéines sécrétées et leurs utilisations

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200680041301.2

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 2619455

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2008540237

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2006315686

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 3927/DELNP/2008

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 1020087011171

Country of ref document: KR

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2006315686

Country of ref document: AU

Date of ref document: 20061113

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2006837407

Country of ref document: EP